1
|
Zhang Y, Lv X, Chen L, Liu Y. The role and function of CLU in cancer biology and therapy. Clin Exp Med 2023; 23:1375-1391. [PMID: 36098834 DOI: 10.1007/s10238-022-00885-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/29/2022] [Indexed: 11/03/2022]
Abstract
Clusterin (CLU) is a highly evolutionary conserved glycoprotein with multiple isoform-specific functions and is widely distributed in different species. Accumulated evidence has shown the prominent role of CLU in regulating several essential physiological processes, including programmed cell death, metastasis, invasion, proliferation and cell growth via regulating diverse signaling pathways to mediate cancer progression in various cancers, such as prostate, breast, lung, liver, colon, bladder and pancreatic cancer. Several studies have revealed the potential benefit of inhibiting CLU in CLU inhibition-based targeted cancer therapies in vitro, in vivo or in human, suggesting CLU is a promising therapeutic target. This review discusses the multiple functions and mechanisms of CLU in regulating tumor progression of various cancers and summarizes the inhibitors of CLU used in CLU inhibition-based targeted cancer therapies.
Collapse
Affiliation(s)
- Yefei Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Department of Biochemistry, Institute of Cancer, College of Life Science, Nanjing Normal University, Nanjing, 210023, People's Republic of China
| | - Xiang Lv
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Department of Biochemistry, Institute of Cancer, College of Life Science, Nanjing Normal University, Nanjing, 210023, People's Republic of China
| | - Liming Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Department of Biochemistry, Institute of Cancer, College of Life Science, Nanjing Normal University, Nanjing, 210023, People's Republic of China.
| | - Yan Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Department of Biochemistry, Institute of Cancer, College of Life Science, Nanjing Normal University, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
2
|
Milosevic B, Stojanovic B, Cvetkovic A, Jovanovic I, Spasic M, Stojanovic MD, Stankovic V, Sekulic M, Stojanovic BS, Zdravkovic N, Mitrovic M, Stojanovic J, Laketic D, Vulovic M, Cvetkovic D. The Enigma of Mammaglobin: Redefining the Biomarker Paradigm in Breast Carcinoma. Int J Mol Sci 2023; 24:13407. [PMID: 37686210 PMCID: PMC10487666 DOI: 10.3390/ijms241713407] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
The continuous evolution of cancer biology has led to the discovery of mammaglobin, a potential novel biomarker for breast carcinoma. This review aims to unravel the enigmatic aspects of mammaglobin and elucidate its potential role in redefining the paradigm of breast carcinoma biomarkers. We will thoroughly examine its expression in tumoral and peritumoral tissues and its circulating levels in the blood, thereby providing insights into its possible function in cancer progression and metastasis. Furthermore, the potential application of mammaglobin as a non-invasive diagnostic tool and a target for personalized treatment strategies will be discussed. Given the increasing incidence of breast carcinoma worldwide, the exploration of novel biomarkers such as mammaglobin is crucial in advancing our diagnostic capabilities and treatment modalities, ultimately contributing to improved patient outcomes.
Collapse
Affiliation(s)
- Bojan Milosevic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.M.); (B.S.); (A.C.)
| | - Bojan Stojanovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.M.); (B.S.); (A.C.)
| | - Aleksandar Cvetkovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.M.); (B.S.); (A.C.)
| | - Ivan Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Marko Spasic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.M.); (B.S.); (A.C.)
| | - Milica Dimitrijevic Stojanovic
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.D.S.); (V.S.)
| | - Vesna Stankovic
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.D.S.); (V.S.)
| | - Marija Sekulic
- Department of Hygiene and Ecology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Bojana S. Stojanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Natasa Zdravkovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Minja Mitrovic
- Department of Neurology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Jasmina Stojanovic
- Department of Otorhinolaryngology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Darko Laketic
- Institute of Anatomy, Faculty of Medicine, University of Belgrade,11000 Belgrade, Serbia;
| | - Maja Vulovic
- Department of Anatomy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Danijela Cvetkovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| |
Collapse
|
3
|
Shi W, Chen S, Chi F, Qiu Q, Zhong Y, Bian X, Zhang H, Xi J, Qian H. Advances in Tumor Antigen‐Based Anticancer Immunotherapy: Recent Progress, Prevailing Challenges, and Future Perspective. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Wei Shi
- Center of Drug Discovery State Key Laboratory of Natural Medicines China Pharmaceutical University 24 Tongjiaxiang Nanjing 210009 P. R. China
| | - Shuang Chen
- Center of Drug Discovery State Key Laboratory of Natural Medicines China Pharmaceutical University 24 Tongjiaxiang Nanjing 210009 P. R. China
| | - Fanglian Chi
- Center of Drug Discovery State Key Laboratory of Natural Medicines China Pharmaceutical University 24 Tongjiaxiang Nanjing 210009 P. R. China
| | - Qianqian Qiu
- Center of Drug Discovery State Key Laboratory of Natural Medicines China Pharmaceutical University 24 Tongjiaxiang Nanjing 210009 P. R. China
| | - Yue Zhong
- Center of Drug Discovery State Key Laboratory of Natural Medicines China Pharmaceutical University 24 Tongjiaxiang Nanjing 210009 P. R. China
| | - Xiaojian Bian
- Center of Drug Discovery State Key Laboratory of Natural Medicines China Pharmaceutical University 24 Tongjiaxiang Nanjing 210009 P. R. China
| | - Hao Zhang
- School of Science China Pharmaceutical University 24 Tongjiaxiang Nanjing 210009 P. R. China
| | - Junting Xi
- School of Science China Pharmaceutical University 24 Tongjiaxiang Nanjing 210009 P. R. China
| | - Hai Qian
- Center of Drug Discovery State Key Laboratory of Natural Medicines China Pharmaceutical University 24 Tongjiaxiang Nanjing 210009 P. R. China
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease China Pharmaceutical University 24 Tongjiaxiang Nanjing 210009 P. R. China
| |
Collapse
|
4
|
Zachariah NN, Basu A, Gautam N, Ramamoorthi G, Kodumudi KN, Kumar NB, Loftus L, Czerniecki BJ. Intercepting Premalignant, Preinvasive Breast Lesions Through Vaccination. Front Immunol 2021; 12:786286. [PMID: 34899753 PMCID: PMC8652247 DOI: 10.3389/fimmu.2021.786286] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) prevention remains the ultimate cost-effective method to reduce the global burden of invasive breast cancer (IBC). To date, surgery and chemoprevention remain the main risk-reducing modalities for those with hereditary cancer syndromes, as well as high-risk non-hereditary breast lesions such as ADH, ALH, or LCIS. Ductal carcinoma in situ (DCIS) is a preinvasive malignant lesion of the breast that closely mirrors IBC and, if left untreated, develops into IBC in up to 50% of lesions. Certain high-risk patients with DCIS may have a 25% risk of developing recurrent DCIS or IBC, even after surgical resection. The development of breast cancer elicits a strong immune response, which brings to prominence the numerous advantages associated with immune-based cancer prevention over drug-based chemoprevention, supported by the success of dendritic cell vaccines targeting HER2-expressing BC. Vaccination against BC to prevent or interrupt the process of BC development remains elusive but is a viable option. Vaccination to intercept preinvasive or premalignant breast conditions may be possible by interrupting the expression pattern of various oncodrivers. Growth factors may also function as potential immune targets to prevent breast cancer progression. Furthermore, neoantigens also serve as effective targets for interception by virtue of strong immunogenicity. It is noteworthy that the immune response also needs to be strong enough to result in target lesion elimination to avoid immunoediting as it may occur in IBC arising from DCIS. Overall, if the issue of vaccine targets can be solved by interrupting premalignant lesions, there is a potential to prevent the development of IBC.
Collapse
Affiliation(s)
| | - Amrita Basu
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Namrata Gautam
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Ganesan Ramamoorthi
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Krithika N Kodumudi
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Nagi B Kumar
- Clinical Science Division, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Loretta Loftus
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Brian J Czerniecki
- Department of Breast Surgery, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
5
|
Abstract
We characterized the proteome profile of mid-lactation small-tailed Han (STH) and DairyMeade (DM) ovine milk in order to explore physiological variation and differences in milk traits between the two breeds. Methodology combined a tandem mass tag (TMT) proteomic approach with LC-MS/MS technology. A total of 656 proteins were identified in STH and DM ovine milk, of which 17and 29 proteins were significantly upregulated (P < 0.05) in STH and DM, respectively. Immune-related proteins and disease-related proteins were highly expressed in STH milk, whereas S100A2 and AEBP1 were highly expressed in DM milk, which had beneficial effects on mammary gland development and milk yield. Our results provide a theoretical basis for future breeding of dairy sheep.
Collapse
|
6
|
Fracol M, Shah N, Dolivo D, Hong S, Giragosian L, Galiano R, Mustoe T, Kim JYS. Can Breast Implants Induce Breast Cancer Immunosurveillance? An Analysis of Antibody Response to Breast Cancer Antigen following Implant Placement. Plast Reconstr Surg 2021; 148:287-298. [PMID: 34398081 DOI: 10.1097/prs.0000000000008165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Women with cosmetic breast implants have significantly lower rates of subsequent breast cancer than the general population (relative risk, 0.63; 95 percent CI, 0.56 to 0.71). The authors hypothesize that breast implant-induced local inflammation stimulates immunosurveillance recognition of breast tumor antigen. METHODS Sera were collected from two cohorts of healthy women: women with long-term breast implants (i.e., breast implants for >6 months) and breast implant-naive women. Antibody responses to breast tumor antigens were tested by enzyme-linked immunosorbent assay and compared between cohorts by unpaired t test. Of the implant-naive cohort, nine women underwent breast augmentation, and antibody responses before and after implant placement were compared by paired t test. RESULTS Sera were collected from 104 women: 36 (34.6 percent) long-term breast implants and 68 (65.4 percent) implant-naive women. Women with long-term breast implants had higher antibody responses than implant-naive women to mammaglobin-A (optical density at 450 nm, 0.33 versus 0.22; p = 0.003) and mucin-1 (optical density at 450 nm, 0.42 versus 0.34; p = 0.02). There was no difference in antibody responses to breast cancer susceptibility gene 2, carcinoembryonic antigen, human epidermal growth factor receptor-2, or tetanus. Nine women with longitudinal samples preoperatively and 1 month postoperatively demonstrated significantly elevated antibody responses following implant placement to mammaglobin-A (mean difference, 0.13; p = 0.0002) and mucin-1 (mean difference 0.08; p = 0.02). There was no difference in postimplant responses to other breast tumor antigens, or tetanus. CONCLUSIONS Women with long-term breast implants have higher antibody recognition of mammaglobin-A and mucin-1. This study provides the first evidence of implant-related immune responses to breast cancer antigens. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, V.
Collapse
Affiliation(s)
- Megan Fracol
- From the Division of Plastic and Reconstructive Surgery, Northwestern University Feinberg School of Medicine
| | - Nikita Shah
- From the Division of Plastic and Reconstructive Surgery, Northwestern University Feinberg School of Medicine
| | - David Dolivo
- From the Division of Plastic and Reconstructive Surgery, Northwestern University Feinberg School of Medicine
| | - Seok Hong
- From the Division of Plastic and Reconstructive Surgery, Northwestern University Feinberg School of Medicine
| | - Lexa Giragosian
- From the Division of Plastic and Reconstructive Surgery, Northwestern University Feinberg School of Medicine
| | - Robert Galiano
- From the Division of Plastic and Reconstructive Surgery, Northwestern University Feinberg School of Medicine
| | - Thomas Mustoe
- From the Division of Plastic and Reconstructive Surgery, Northwestern University Feinberg School of Medicine
| | - John Y S Kim
- From the Division of Plastic and Reconstructive Surgery, Northwestern University Feinberg School of Medicine
| |
Collapse
|
7
|
Kar SP, Considine DP, Tyrer JP, Plummer JT, Chen S, Dezem FS, Barbeira AN, Rajagopal PS, Rosenow WT, Moreno F, Bodelon C, Chang-Claude J, Chenevix-Trench G, deFazio A, Dörk T, Ekici AB, Ewing A, Fountzilas G, Goode EL, Hartman M, Heitz F, Hillemanns P, Høgdall E, Høgdall CK, Huzarski T, Jensen A, Karlan BY, Khusnutdinova E, Kiemeney LA, Kjaer SK, Klapdor R, Köbel M, Li J, Liebrich C, May T, Olsson H, Permuth JB, Peterlongo P, Radice P, Ramus SJ, Riggan MJ, Risch HA, Saloustros E, Simard J, Szafron LM, Titus L, Thompson CL, Vierkant RA, Winham SJ, Zheng W, Doherty JA, Berchuck A, Lawrenson K, Im HK, Manichaikul AW, Pharoah PD, Gayther SA, Schildkraut JM. Pleiotropy-guided transcriptome imputation from normal and tumor tissues identifies candidate susceptibility genes for breast and ovarian cancer. HGG ADVANCES 2021; 2:100042. [PMID: 34317694 PMCID: PMC8312632 DOI: 10.1016/j.xhgg.2021.100042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
Familial, sequencing, and genome-wide association studies (GWASs) and genetic correlation analyses have progressively unraveled the shared or pleiotropic germline genetics of breast and ovarian cancer. In this study, we aimed to leverage this shared germline genetics to improve the power of transcriptome-wide association studies (TWASs) to identify candidate breast cancer and ovarian cancer susceptibility genes. We built gene expression prediction models using the PrediXcan method in 681 breast and 295 ovarian tumors from The Cancer Genome Atlas and 211 breast and 99 ovarian normal tissue samples from the Genotype-Tissue Expression project and integrated these with GWAS meta-analysis data from the Breast Cancer Association Consortium (122,977 cases/105,974 controls) and the Ovarian Cancer Association Consortium (22,406 cases/40,941 controls). The integration was achieved through application of a pleiotropy-guided conditional/conjunction false discovery rate (FDR) approach in the setting of a TWASs. This identified 14 candidate breast cancer susceptibility genes spanning 11 genomic regions and 8 candidate ovarian cancer susceptibility genes spanning 5 genomic regions at conjunction FDR < 0.05 that were >1 Mb away from known breast and/or ovarian cancer susceptibility loci. We also identified 38 candidate breast cancer susceptibility genes and 17 candidate ovarian cancer susceptibility genes at conjunction FDR < 0.05 at known breast and/or ovarian susceptibility loci. The 22 genes identified by our cross-cancer analysis represent promising candidates that further elucidate the role of the transcriptome in mediating germline breast and ovarian cancer risk.
Collapse
Affiliation(s)
- Siddhartha P. Kar
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Daniel P.C. Considine
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Jonathan P. Tyrer
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Jasmine T. Plummer
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Stephanie Chen
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Felipe S. Dezem
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Alvaro N. Barbeira
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Padma S. Rajagopal
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Will T. Rosenow
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Fernando Moreno
- Department of Oncology, Hospital Clínico San Carlos, Madrid, Spain
| | - Clara Bodelon
- Divison of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Cancer Epidemiology Group, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Georgia Chenevix-Trench
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Anna deFazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
| | - Thilo Dörk
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Arif B. Ekici
- Institute of Human Genetics, University Hospital Erlangen, Erlangen, Germany
- Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen, Erlangen, Germany
| | - Ailith Ewing
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Ellen L. Goode
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Mikael Hartman
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Florian Heitz
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte/Evang., Essen, Germany
- Department of Gynecology, Center for Oncologic Surgery, Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Peter Hillemanns
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Estrid Høgdall
- Department of Virus, Lifestyle, and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
- Molecular Unit, Department of Pathology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Claus K. Høgdall
- The Juliane Marie Centre, Department of Gynecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Tomasz Huzarski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
- Department of Genetics and Pathology, University of Zielona Góra, Zielona Góra, Poland
| | - Allan Jensen
- Department of Lifestyle, Reproduction, and Cancer, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Beth Y. Karlan
- David Geffen School of Medicine, Department of Obstetrics and Gynecology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Elza Khusnutdinova
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia
- Department of Genetics and Fundamental Medicine, Bashkir State University, Ufa, Russia
| | - Lambertus A. Kiemeney
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Susanne K. Kjaer
- Department of Virus, Lifestyle, and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Rüdiger Klapdor
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, Calgary, AB, Canada
| | - Jingmei Li
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
- Genome Institute of Singapore, Human Genetics, Singapore, Singapore
| | - Clemens Liebrich
- Department of Obstetrics and Gynecology, Klinikum Wolfsburg, Wolfsburg, Germany
| | - Taymaa May
- Division of Gynecologic Oncology, University Health Network, Princess Margaret Hospital, Toronto, ON, Canada
| | - Håkan Olsson
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jennifer B. Permuth
- Departments of Cancer Epidemiology and Gastrointestinal Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Paolo Peterlongo
- Genome Diagnostics Program, IFOM-The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Paolo Radice
- Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori (INT), Milan, Italy
| | - Susan J. Ramus
- School of Women’s and Children’s Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Adult Cancer Program, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Marjorie J. Riggan
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Harvey A. Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | | | - Jacques Simard
- Genomics Center, Centre Hospitalier Universitaire de Québec - Université Laval Research Center, Québec City, QC, Canada
| | - Lukasz M. Szafron
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Linda Titus
- Muskie School of Public Service, University of Southern Maine, Portland, ME, USA
| | - Cheryl L. Thompson
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - Robert A. Vierkant
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Stacey J. Winham
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jennifer A. Doherty
- Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Andrew Berchuck
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Kate Lawrenson
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Women’s Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hae Kyung Im
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Ani W. Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Paul D.P. Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Simon A. Gayther
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joellen M. Schildkraut
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| |
Collapse
|
8
|
Oloomi M, Moazzezy N, Bouzari S. Comparing blood versus tissue-based biomarkers expression in breast cancer patients. Heliyon 2020; 6:e03728. [PMID: 32274439 PMCID: PMC7132155 DOI: 10.1016/j.heliyon.2020.e03728] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 01/14/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022] Open
Abstract
Molecular markers have been used as a tool for diagnostic approaches, staging, and evaluation of therapeutic responses in patients with cancer. Cancer molecular markers can also help clinicians to make decision on therapy and prognosis evaluation at the time of diagnosis. In the early diagnosis of breast cancer (BC), estrogen and progesterone receptors (ER/PR) expression levels should be determined through immunohistochemistry (IHC). In molecular genetics, there are some important tissue-based markers that can also be found in blood, such as Mammaglobin (MAM), Cytokeratin 19 (CK19), Mucin (MUC), Proto-oncogene (c-Myc), antigen Ki-67 (Ki67), and Carcinoembryonic antigen (CEA). In this study, the positive level of the marker genes in both blood and tissue of the BC patients were compared using reverse transcription polymerase chain reaction (RT-PCR) method. In addition, the importance of blood vs. tissue-based markers in BC diagnosis also demonstrated and is discussed. CEA (O), ERβ, CK19 and, c-Myc molecular markers were significantly different between blood of normal and patients while there was no significant difference of these markers in tissue samples. Blood-based biomarkers can be used for the early diagnosis of BC. Comparing blood versus tissue-based biomarkers indicated that there are correlations between markers in blood and tissue, since blood markers can be substitute to tissue markers in BC patients in the future.
Collapse
Affiliation(s)
- Mana Oloomi
- Molecular Biology Department, Pasteur Institute of Iran, Pasteur Ave., Tehran, 13164, Iran
| | | | | |
Collapse
|
9
|
Attenuated Total Reflection-Fourier Transform Infrared (ATR-FTIR) Spectroscopy Analysis of Saliva for Breast Cancer Diagnosis. JOURNAL OF ONCOLOGY 2020; 2020:4343590. [PMID: 32104176 PMCID: PMC7035572 DOI: 10.1155/2020/4343590] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/28/2019] [Indexed: 12/15/2022]
Abstract
Saliva biomarkers using reagent-free biophotonic technology have not been investigated as a strategy for early detection of breast cancer (BC). The attenuated total reflection-Fourier transform infrared (ATR-FTIR) spectroscopy has been proposed as a promising tool for disease diagnosis. However, its utilization in cancer is still incipient, and currently saliva has not been used for BC screening. We have applied ATR-FTIR onto saliva from patients with breast cancer, benign breast disease, and healthy matched controls to investigate its potential use in BC diagnosis. Several salivary vibrational modes have been identified in original and second-derivative spectra. The absorbance levels at wavenumber 1041 cm−1 were significantly higher (p < 0.05) in saliva of breast cancer patients compared with those of benign patients, and the ROC curve analysis of this peak showed a reasonable accuracy to discriminate breast cancer from benign and control patients. The 1433–1302.9 cm−1 band area was significantly higher (p < 0.05) in saliva of breast cancer patients than in control and benign patients. This salivary ATR-FTIR spectral area was prevalidated as a potential diagnostic biomarker of BC. This spectral biomarker was able to discriminate human BC from controls with sensitivity and specificity of 90% and 80%, respectively. Besides, it was able to differentiate BC from benign disease with sensitivity and specificity of 90% and 70%, respectively. Briefly, for the first time, saliva analysis by ATR-FTIR spectroscopy has demonstrated the potential use of salivary spectral biomarkers (1041 cm−1 and 1433–1302.9 cm−1) as a novel alternative for noninvasive BC diagnosis, which could be used for screening purposes.
Collapse
|
10
|
Macron C, Lane L, Núñez Galindo A, Dayon L. Deep Dive on the Proteome of Human Cerebrospinal Fluid: A Valuable Data Resource for Biomarker Discovery and Missing Protein Identification. J Proteome Res 2018; 17:4113-4126. [PMID: 30124047 DOI: 10.1021/acs.jproteome.8b00300] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Cerebrospinal fluid (CSF) is a body fluid of choice for biomarker studies of brain disorders but remains relatively under-studied compared with other biological fluids such as plasma, partly due to the more invasive means of its sample collection. The present study establishes an in-depth CSF proteome through the analysis of a unique CSF sample from a pool of donors. After immunoaffinity depletion, the CSF sample was fractionated using off-gel electrophoresis and analyzed with liquid chromatography tandem mass spectrometry (MS) using the latest generation of hybrid Orbitrap mass spectrometers. The shotgun proteomic analysis allowed the identification of 20 689 peptides mapping on 3379 proteins. To the best of our knowledge, the obtained data set constitutes the largest CSF proteome published so far. Among the CSF proteins identified, 34% correspond to genes whose transcripts are highly expressed in brain according to the Human Protein Atlas. The principal Alzheimer's disease biomarkers (e.g., tau protein, amyloid-β, apolipoprotein E, and neurogranin) were detected. Importantly, our data set significantly contributes to the Chromosome-centric Human Proteome Project (C-HPP), and 12 proteins considered as missing are proposed for validation in accordance with the HPP guidelines. Of these 12 proteins, 8 proteins are based on 2 to 6 uniquely mapping peptides from this CSF analysis, and 4 match a new peptide with a "stranded" single peptide in PeptideAtlas from previous CSF studies. The MS proteomic data are available to the ProteomeXchange Consortium ( http://www.proteomexchange.org/ ) with the data set identifier PXD009646.
Collapse
Affiliation(s)
- Charlotte Macron
- Proteomics , Nestlé Institute of Health Sciences , 1015 Lausanne , Switzerland
| | - Lydie Lane
- CALIPHO Group , SIB-Swiss Institute of Bioinformatics , CMU, rue Michel-Servet 1 , 1211 Geneva 4 , Switzerland.,Department of Microbiology and Molecular Medicine, Faculty of Medicine , University of Geneva , rue Michel-Servet 1 , 1211 Geneva 4 , Switzerland
| | | | - Loïc Dayon
- Proteomics , Nestlé Institute of Health Sciences , 1015 Lausanne , Switzerland
| |
Collapse
|
11
|
Anti-hMAM monoclonal antibodies evaluated in breast and non-breast tissues for differential diagnosis implication. TUMORI JOURNAL 2016; 2016:264-9. [PMID: 27103207 DOI: 10.5301/tj.5000496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2016] [Indexed: 11/20/2022]
Abstract
PURPOSE Human mammaglobin (hMAM) is a breast tissue-specific marker that may have potency for the diagnosis of breast cancer. However, there is a lack of commercialization of anti-hMAM antibody made in China. METHODS Immunoreactivities of 2 self-made monoclonal anti-hMAM antibodies, MEF521 and MDA822, were evaluated by immunohistochemistry staining and compared with imported monoclonal antibody ab81611. A total of 48 cases of primary breast cancers, 36 cases of benign or normal breast tissues, 52 cases of lymph nodes or organ metastases from breast cancer, and 90 cases of non-breast primary carcinoma tissues were analyzed. RESULTS All 3 anti-hMAM antibodies showed high positive expression of hMAM in primary breast cancers, benign, and normal breast tissues. The positive ratio for MEF521 (33.3%) or MDA822 (44.4%) was much higher than that of ab81611 (16.7%) in lymph node metastasis from breast cancer (p = 0.038). There was no correlation between hMAM expression and clinicopathologic features of breast cancer in the 3 groups of antibodies. In 90 cases of non-breast primary carcinoma tissues, no hMAM-positive ones were observed in the MEF521 or MDA822 group, but 48 (53.3%) in the ab81611 group were positive, indicating that breast tissue specificity of the 2 self-made anti-hMAM monoclonal antibodies much higher than that of ab81611 (p<0.001). CONCLUSIONS Our results showed that MDA822 and MEF521 are more specific to breast cancer as measured by means of immunohistochemistry. Therefore, the 2 self-made anti-hMAM antibodies may have good prospects for clinical application in the differential diagnosis of breast tumor and breast cancer metastases.
Collapse
|
12
|
Review: circulating tumor cells in the practice of breast cancer oncology. Clin Transl Oncol 2015; 18:749-59. [DOI: 10.1007/s12094-015-1460-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022]
|
13
|
Duan C, Yang X, Zhang X, Feng J, Liu Z, Que H, Johnson H, Zhao Y, Fan Y, Lu Y, Zhang H, Huang Y, Xiu B, Feng X. Generation of monoclonal antibodies against MGA and comparison of their application in breast cancer detection by immunohistochemistry. Sci Rep 2015; 5:13073. [PMID: 26272389 PMCID: PMC4536492 DOI: 10.1038/srep13073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 07/16/2015] [Indexed: 11/15/2022] Open
Abstract
Mammaglobin A (MGA) is an organ specific molecular biomarker for metastatic breast cancer diagnosis. However, there are still needs to develop optimal monoclonal antibodies (mAbs) to detect MGA expression in breast carcinoma by immunohistochemistry. In this study, we first generated mAbs against MGA. Then, we used epitope prediction and computer-assisted structural analysis to screen five dominant epitopes and identified mAbs against five epitopes. Further immunohistochemical analysis on 42 breast carcinoma specimens showed that MHG1152 and MGD785 had intensive staining mainly in membrane, while CHH11617, CHH995 and MJF656 had more intensive staining within the cytoplasm. MGA scoring results showed that MJF656 had the highest rate (92.8%) of positive staining among five mAbs, including higher staining intensity when compared with that of MHG1152 (p < 0.01) and CHH995 (p < 0.05) and the highest the mean percentage of cells stained among mAbs. Furthermore, we analyzed the relationship of positive staining rate by mAbs with patient clinical characteristics. The results suggest that MJF656 was able to detect MGA expression, especially in early clinical stage, low grade and lymph node metastasis-negative breast carcinoma. In conclusion, our study generated five mAbs against MGA and identified the best candidate for detection of MGA expression in breast cancer tissues.
Collapse
Affiliation(s)
- Cuimi Duan
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Xiqin Yang
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Xuhui Zhang
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Jiannan Feng
- Department of Immunology, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Zhiqiang Liu
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Haiping Que
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | | | - Yanfeng Zhao
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Yawen Fan
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Yinglin Lu
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Heqiu Zhang
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Yan Huang
- Affiliated 307 Hospital, Beijing, 100071, China
| | - Bingshui Xiu
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Xiaoyan Feng
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| |
Collapse
|